Anemia of Chronic Diseases

NV Kurkina, EI Gorshenina, LV Chegodaeva, AV Polagimova

NP Ogarev National Research Mordovia State University, 68 Bolshevistskaya str., Saransk, Russian Federation, 430005

For correspondence: Nadezhda Viktorovna Kurkina, MD, PhD, 26А Ul’yanova str., Saransk, Russian Federation, 430032; Tel.: +7(927)172-48-63; e-mail: nadya.kurckina@yandex.ru

For citation: Kurkina NV, Gorshenina EI, Chegodaeva LV, Polagimova AV. Anemia of Chronic Diseases. Clinical oncohematology. 2021;14(3):347–54. (In Russ).

DOI: 10.21320/2500-2139-2021-14-3-347-354


ABSTRACT

Anemia of chronic diseases (ACD) refers to a group of anemias arising in various inflammatory infections, autoimmune or tumor diseases due to acute or chronic immune activation. ACD ranks second in incidence after iron deficiency anemia (IDA). Within the variety of pathogenetic mechanisms one of the primary ones is hepcidin synthesis in hepatocytes, which blocks iron absorption in the intestine and contributes to its deposition in cells of the monocyte-macrophage system. Besides, excessive cytokines in such diseases and pathologies lead to lower erythropoietin production which does not correspond to the severity grade of anemia. This results in impaired erythropoiesis in the bone marrow. The differential diagnosis should also specify iron deficiency type (the absolute one in IDA and the functional one in ACD). The effective treatment of the main disease and anemia correction speed up the improvement of patient’s status, rehabilitation, and quality of life.

Keywords: anemia, chronic diseases, immune system, hepcidin, cytokines, erythropoietin, ferritin, serum iron.

Received: January 17, 2021

Accepted: April 30, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Андреичев Н.А., Балеева Л.В. Анемия хронических заболеваний. Российский медицинский журнал. 2014;20(2):50–5.
    [Andreichev NA, Baleeva LV. Anemia of chronic diseases. Rossiiskii meditsinskii zhurnal. 2014;20(2):50–5. (In Russ)]
  2. Волкова С.А., Боровков Н.Н. Основы клинической гематологии: учебное пособие. Н. Новгород: НижГМА, 2013. 400 с.
    [Volkova SA, Borovkov NN. Osnovy klinicheskoi gematologii: uchebnoe posobie. (Fundamentals of clinical hematology: learning guide.) Nizhny Novgorod: NizhGMA Publ.; 2013. 400 p. (In Russ)]
  3. John M, Hoernig S, Doehner W, et al. Anemia and inflammation in COPD. Chest. 2005;127(3):825–9. doi: 10.1378/chest.127.3.825.
  4. Будневский А.В., Есауленко И.Е., Овсянников Е.С., Жусина Ю.Г. Анемия при хронической обструктивной болезни легких. Терапевтический архив. 2016;88(3):96–9. doi: 10.17116/terarkh201688396-99.
    [Budnevsky AV, Esaulenko IE, Ovsyannikov ES, Zhusina YuG. Anemia in chronic obstructive pulmonary disease. Terapevticheskii arkhiv. 2016;88(3):96–9. doi: 10.17116/terarkh201688396-99. (In Russ)]
  5. Жусина Ю.Г., Будневский А.В., Феськова А.А., Овсянников Е.С. О взаимосвязи хронической обструктивной болезни легких и анемии. Пульмонология. 2018;28(6):730–5. doi: 10.18093/0869-0189-2018-28-6-730-735.
    [Zhusina YuG, Budnevskiy AV, Fes’kova AA, Ovsyannikov ES. About relationship between chronic obstructive pulmonary disease and anemia. Pulmonologiya. 2018;28(6):730–5. doi: 10.18093/0869-0189-2018-28-6-730-735. (In Russ)]
  6. Tsantes AE, Tassiopoulos ST, Papadhimitriou SI, et al. Theophylline treatment may adversely affect the anoxia-induced erythropoietic response without suppressing erythropoietin production. Eur J Clin Pharmacol. 2003;59(5–6):379–83. doi: 10.1007/s00228-003-0640-0.
  7. Marathias KP, Agroyannis B, Mavromoustakos T, et al. Hematocrit-lowering effect following inactivation of renin-angiotensin system with angiotensin converting enzyme inhibitors and angiotensin receptor blockers. Curr Top Med Chem. 2004;4(4):483–6. doi: 10.2174/1568026043451311.
  8. Рукавицын О.А. Гематология. Национальное руководство. М.: ГЭОТАР-Медиа, 2017. 784 с.
    [Rukavitsyn OA. Natsional’noe rukovodstvo. (Hematology. National Guidelines.) Moscow: GEOTAR-Media Publ.; 2017. 784 p. (In Russ)]
  9. Groenveld HF, Januzzi JL, Damman K, et al. Anemia and mortality in heart failure patients a systematic review and meta-analysis. J Am Coll Cardiol. 2008;52(10):818–27. doi: 10.1016/j.jacc.2008.04.061.
  10. Снеговой А.В., Aapro M., Гладков О.А. и др. Практические рекомендации по лечению анемии у онкологических больных. Злокачественные опухоли. 2016;4:368–77.
    [Snegovoi AV, Aapro M, Gladkov OA, et al. Practical guidelines for anemia treatment in oncological patients. Zlokachestvennye opukholi. 2016;4:368–77. (In Russ)]
  11. Voulgari PV, Kolios G, Papadopoulos GK, et al. Role of cytokines in the pathogenesis of anemia of chronic disease in rheumatoid arthritis. Clin Immunol. 1999;92(2):153–60. doi: 10.1006/clim.1999.4736.
  12. Stauffer ME, Fan T. Prevalence of Anemia in Chronic Kidney Disease in the United States. PLoS One. 2014;9(1):e84943. doi: 10.1371/journal.pone.0084943.
  13. McClellan W, Aronoff SL, Bolton WK, et al. The prevalence of anemia in patients with chronic kidney disease. Curr Med Ress Opion. 2004;20(9):1501–10. doi: 10.1185/030079904X2763.
  14. Stenvinkel P. The role of inflammation in the anaemia of end-stage renal disease. Nephrol Dial Transplant. 2001;16(Suppl 7):36–40. doi: 10.1093/ndt/16.suppl_7.36.
  15. Thorp ML, Johnson ES. Effect of anemia on mortality, cardiovascular hospitalizations and end stage renal disease among patients with chronic kidney disease. Nephrology. 2009;14(2):240–6. doi: 10.1111/j.1440-1797.2008.01065.x.
  16. Andrews M, Arredondo M. Ferritin levels and hepcidin mRNA expression in peripheral mononuclear cells from anemic type 2 diabetic patients. Biol Trace Elem Res. 2012;149(1):1–4. doi: 10.1007/s12011-012-9389-6.
  17. Zoppini G, Targher G, Chonchol M, et al. Anaemia, independent of chronic kidney disease, predicts all cause and cardiovascular mortality in type 2 diabetic patients. Atherosclerosis. 2010;210(2):575–80. doi: 10.1016/j.atherosclerosis.2009.12.008.
  18. Ito H, Takeuchi Y, Ishida H, et al. Mild anemia is frequent and associated with micro- and macroangiopathies in patients with type 2 diabetes mellitus. J Diab Invest. 2010;1(6):273–8. doi: 10.1111/j.2040-1124.2010.00060.x.
  19. Roy CN, Mak HH, Akpan I, et al. Hepcidin antimicrobial peptide transgenic mice exhibit features of the anemia of inflammation. Blood. 2007;109(9):4038–44. doi: 10.1182/blood-2006-10-051755.
  20. Ganz T, Nemeth E. Iron sequestration and anemia of inflammation. Semin Hematol. 2009;46(4):387–393. doi: 10.1053/j.seminhematol.2009.06.001.
  21. Морщакова Е.Ф., Павлов А.Д., Румянцев А.Г. Эритропоэз, эритропоэтин, железо. М.: ГЭОТАР-Медиа, 2013. 178 с.
    [Morshchakova EF, Pavlov AD, Rumyantsev AG. Eritropoez, eritropoetin, zhelezo. (Erythropoiesis, erythropoietin, iron.) Moscow: GEOTAR-Media Publ.; 2013. 178 p. (In Russ)]
  22. Рукавицын О.А. Анемия хронических заболеваний: отдельные аспекты патогенеза и пути коррекции. Онкогематология. 2016;11(1):37–46. doi: 10.17650/1818-8346-2016-11-1-37-46.
    [Rukavitsyn OA. Anemia of chronic diseases: the important aspects of pathogenesis and treatment. Oncohematology. 2016;11(1):37–46. doi: 10.17650/1818-8346-2016-11-1-37-46. (In Russ)]
  23. Румянцев А.Г., Масчан А.А. Федеральные клинические рекомендации по диагностике и лечению анемии хронических заболеваний (электронный документ). Доступно по: https://nodgo.org/sites/default/files/%D0%A4%D0%9A%D0%A0%20%D0%BF%D0%BE%20%D0%B4%D0%B8%D0%B0%D0%B3%D0%BD%D0%BE%D1%81%D1%82%D0%B8%D0%BA%D0%B5%20%D0%B8%20%D0%BB%D0%B5%D1%87%D0%B5%D0%BD%D0%B8%D1%8E%20%D0%B0%D0%BD%D0%B5%D0%BC%D0%B8%D0%B8%20%D1%85%D1%80%D0%BE%D0%BD%D0%B8%D1%87%D0%B5%D1%81%D0%BA%D0%B8%D1%85%20%D0%B1%D0%BE%D0%BB%D0%B5%D0%B7%D0%BD%D0%B5%D0%Bpdf. Ссылка активна на 13.04.2021.
    [Rumyantsev AG, Maschan AA. Federal clinical guidelines for diagnosis and treatment of anemia of chronic diseases. [Internet] Available from: https://nodgo.org/sites/default/files/%D0%A4%D0%9A%D0%A0%20%D0%BF%D0%BE%20%D0%B4%D0%B8%D0%B0%D0%B3%D0%BD%D0%BE%D1%81%D1%82%D0%B8%D0%BA%D0%B5%20%D0%B8%20%D0%BB%D0%B5%D1%87%D0%B5%D0%BD%D0%B8%D1%8E%20%D0%B0%D0%BD%D0%B5%D0%BC%D0%B8%D0%B8%20%D1%85%D1%80%D0%BE%D0%BD%D0%B8%D1%87%D0%B5%D1%81%D0%BA%D0%B8%D1%85%20%D0%B1%D0%BE%D0%BB%D0%B5%D0%B7%D0%BD%D0%B5%D0%B9.pdf. (accessed 13.04.2021) (In Russ)]
  24. Nemeth E, Ganz T. Anemia of Inflammation. Hematol Oncol Clin North Am. 2014;28(4):671–81. doi: 10.1016/j.hoc.2014.04.005.
  25. Weiss Pathogenesis and treatment of anemia of chronic disease. Blood Rev. 2002;16(2):87–96. doi: 10.1054/blre.2002.0193.
  26. Сморкалова Е.В. Иммуногематологические особенности железодефицитной анемии и анемии хронических заболеваний: Автореф. дис.… канд. мед. наук. Уфа, 2012. 22 с.
    [Smorkalova EV. Immunogematologicheskie osobennosti zhelezodefitsitnoi anemii i anemii khronicheskikh zabolevanii. (Immunohematological characteristics of iron deficiency anemia and anemia of chronic diseases.) [dissertation] Ufa; 2012. 22 p. (In Russ)]
  27. Kato Y, Takagi C, Tanaka J, et al. Effect of daily subcutaneous administration of recombinant erythropoietin on chronic anemia in rheumatoid arthritis. Intern Med. 1994;33(4):193–7. doi: 10.2169/internalmedicine.33.193.
  28. Peeters HR, Jongen-Lavrencic M, Bakker CH, et al. Recombinant human erythropoietin improves health-related quality of life in patients with rheumatoid arthritis and anaemia of chronic disease; utility measures correlate strongly with disease activity measures. Rheumatol Int. 1999;18(5–6):201–6. doi: 10.1007/s002960050085.
  29. Arndt U, Kaltwasser JP, Gottschalk R, et al. Correction of iron-deficient erythropoiesis in the treatment of anemia of chronic disease with recombinant human erythropoietin. Ann Hematol. 2005;84(3):159–66. doi: 10.1007/s00277-004-0950-z.
  30. Schipperus M, Rijnbeek B, Reddy M, et al. CNTO328 (Anti-IL-6 mAb) Treatment Is Associated with An Increase in Hemoglobin (Hb) and Decrease in Hepcidin Levels in Renal Cell Carcinoma (RCC). Blood. 2009;114(22):4045. doi: 10.1182/blood.v114.22.4045.4045.
  31. Hohlbaum A, Gille H, Christian J, et al. Iron mobilization and pharmacodynamic marker measurements in non-human primates following administration of PRS-080, a novel and highly specific antihepcidin therapeutic. Am J Hematol. 2013;88(5):E41.
  32. Schwoebel F, van Eijk LT, Zboralski D, et al. The effects of the anti-hepcidin Spiegelmer NOX-H94 on inflammation-induced anemia in cynomolgus monkeys. Blood. 2013;121(12):2311–5. doi: 10.1182/blood-2012-09-456756.
  33. Poli M, Girelli D, Campostrini N, et al. Heparin: a potent inhibitor of hepcidin expression in vitro and in vivo. Blood. 2011;117(3):997–1004. doi: 10.1182/blood-2010-06-289082.
  34. Crosby JR, Gaarde WA, Egerston J, et al. Targeting hepcidin with antisense oligonucleotides improves anemia endpoints in mice. Blood. 2006;108(11, Pt 1):269. doi: 10.1182/blood.v108.11.269.269.
  35. Akinc A, Chan-Daniels A, Sehgal A, et al. Targeting the hepcidin pathway with RNAi therapeutics for the treatment of anemia. Blood. 2011;118(21):688. doi: 10.1182/blood.v118.21.688.688.
  36. Гармиш Е.А. Анемия хронического воспаления при ревматоидном артрите: патогенез и выбор терапии. Украинский ревматологический журнал. 2016;1(63):39–41.
    [Garmish EА. Anemia of chronic inflammation of rheumatoid arthritis: pathogenesis and choice of treatment. Ukrainskii revmatologicheskii zhurnal. 2016;1(63):39–41. (In Russ)]

Anemia of Chronic Disease: Key Mechanisms of Pathogenesis in Patients with Malignancies and Feasible Classification Approaches

VT Sakhin1, ER Madzhanova1, EV Kryukov3, AV Sotnikov2, AV Gordienko2, OA Rukavitsyn3

1 1586 Military Clinical Hospital, 4 Mashtakova str., Moscow Region, Podolsk, Russian Federation, 142110

2 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

3 NN Burdenko Central Military Clinical Hospital, 3 Gospital’naya sq., Moscow, Russian Federation, 105229

For correspondence: Valerii Timofeevich Sakhin, MD, PhD, 4 Mashtakova str., Moscow Region, Podolsk, Russian Federation, 142110; Tel.: +7(916)314-31-11; e-mail: SahinVT@yandex.ru

For citation: Sakhin VT, Madzhanova ER, Kryukov EV, et al. Anemia of Chronic Disease: Key Mechanisms of Pathogenesis in Patients with Malignancies and Feasible Classification Approaches. Clinical oncohematology. 2019;12(3):344–9 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-344-349


ABSTRACT

Aim. To study the effect of hepcidin, soluble transferrin receptor (sTfR), and cytokines on iron metabolism and occurrence of anemia in patients with malignancies and to propose, on this basis, a draft classification of anemia of chronic disease (ACD) based on the major pathogenic factor.

Materials & Methods. The trial included 63 patients with malignancies of stage II/IV: 41 patients with anemia (34 men, 7 women, mean age 67.1 ± 9.9 years), 22 patients without anemia (17 men, 5 women, mean age 60.2 ± 14.9 years). Comparative analysis was based on the values of iron metabolism, C-reactive protein (CRP), hepcidin, sTfR, as well as pro-inflammatory (interleukin-6 [IL-6], tumour necrosis factor α [TNF-α]) and anti-inflammatory (IL-10) cytokines in solid malignancy patients with and without anemia. The correlation analysis between IL-6, IL-10, TNF-α, hepcidin, sTfR, and blood count was performed.

Results. Compared with the control group patients with anemia show lower levels of iron concentration, total iron-binding capacity (TIBC), and percent transferrin saturation (TSAT), as well as higher level of CRP, hepcidin, sTfR, IL-6, IL-10, and TNF-α (< 0.05). IL-6 (r = –0.58), TNF-α (r = –0.32), and hepcidin (r = –0.57) proved to negatively affect erythrocyte level. A negative correlation was established between hemoglobin concentration and IL-6 (r = –0.57), IL-10 (r = –0.64), TNF-α (r = –0.65), hepcidin (r = –0.3), and sTfR (r = –0.57). A correlation was identified between concentrations of hepcidin and IL-6 (r = 0.58), IL-10 (r = 0.33), TNF-α (r = –0.4), as well as between concentrations of sTfR and IL-10 (r = 0.58), TNF-α (r = –0.53). A relationship was identified between IL-6 concentration and iron status (r = –0.38), TIBC (r = –0.56), TSAT (r = –0.31), ferritin (r = 0.56), transferrin (r = –0.72), CRP (r = 0.86) as well as between concentrations of IL-10 and iron (r = –0.63), TSAT (r = –0.67), transferrin (r = –0.7), ferritin (r = 0.55), CRP (r = 0.65), TIBC (r = –0.71). A correlation between the levels of TNF-α and TIBC (r = –0.36), transferrin (r = –0.5) was confirmed.

Conclusion. The paper deals with multi-factorial pathogenesis of anemia in patients with malignancies. Most important factors are iron deficiency and erythropoietic disorder. A draft ACD classification based on the major pathogenic factor of anemia (ACD with dominating iron deficiency, ACD with impaired regulatory mechanism of erythropoiesis, and ACD with insufficient erythropoietin production) is proposed.

Keywords: cancer, anemia, iron metabolism, interleukin-6, interleukin-10, tumor necrosis factor alpha, hepcidin, soluble transferrin receptor.

Received: January 21, 2019

Accepted: June 18, 2019

Read in PDF 


REFERENCES

  1. Weiss G. Pathogenesis and treatment of anaemia of chronic disease. Blood Rev. 2002;16(2):87–96. doi: 10.1054/blre.2002.0193.

  2. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011–23. doi: 10.1056/nejmra041809.

  3. Means RT. Recent developments in the anemia of chronic disease. Curr Hematol Rep. 2003;2(2):116–21.

  4. Poggiali E, De Amicis MM, Motta I, et al. Anemia of chronic disease: a unique defect of iron recycling for many different chronic diseases. Eur J Int Med. 2014;25(1):12–17. doi: 10.1016/j.ejim.2013.07.011.

  5. Weiss G. Iron metabolism in the anemia of chronic disease. Biochim Biophys Acta. 2009;1790(7):682–93. doi: 10.1016/j.bbagen.2008.08.006.

  6. Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys Acta. 2012;1823(9):1434–43. doi: 10.1016/j.bbamcr.2012.01.014.

  7. McCranor BJ, Kim MJ, Cruz NM, et al. Interleukin-6 directly impairs the erythroid development of human TF-1 erythroleukemic cells. Blood Cells Mol Dis. 2014;52(2–3):126–33. doi: 10.1016/j.bcmd.2013.09.004.

  8. Анемии. Под ред. О.А. Рукавицына. 2-е изд., перераб. и доп. М.: ГЭОТАР-Медиа, 2016. 256 c.

    [Rukavitsyn OA, ed. (Anemias.) 2nd revised edition. Moscow: GEOTAR-Media Publ.; 2016. 256 p. (In Russ)]

  9. Сахин В.Т., Маджанова Е.Р., Крюков Е.В. и др. Анемия хронических заболеваний: особенности патогенеза и возможности терапевтической коррекции (обзор литературы и результаты собственных исследований). Онкогематология. 2018;13(1):45–53. doi: 10.17650/1818-8346-2018-13-1-45-53.

    [Sakhin VТ, Madzhanova ЕR, Kryukov EV, et al. Anemia of chronic disease: features of pathogenesis and possible therapeutic correction (literature review and results of own research). Oncohematology. 2018;13(1):45–53. doi: 10.17650/1818-8346-2018-13-1-45-53. (In Russ)]

  10. Steinmetz T, Totzke U, Schweigert M, et al. A prospective observational study of anaemia management in cancer patients–results from the German Cancer Anaemia Registry. Eur J Cancer Care. 2011;20(4):493–502. doi: 10.1111/j.1365-2354.2010.01230.x.

  11. Waters JS, O’Brien MER, Ashley S. Management of anemia in patients receiving chemotherapy. J Clin Oncol. 2002;20(2):601–3. doi: 10.1200/JCO.2002.20.2.601.

  12. Grotto HZ. Anaemia of cancer: an overview of mechanisms involved in its pathogenesis. Med Oncol. 2008;25(1):12–21. doi: 1007/s12032-007-9000-8.

  13. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. С. 143–9.

    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. pp. 143–9. (In Russ)]

  14. Steinmetz HT, Tsamaloukas A, Schmitz S, et al. A new concept for the differential diagnosis and therapy of anaemia in cancer patients. Support Care Cancer. 2010;19(2):261–9. doi: 10.1007/s00520-010-0812-2.

  15. Maccio A, Madeddu C, Massa D, et al. Hemoglobin levels correlate with interleukin-6 levels in patients with advanced untreated epithelial ovarian cancer: role of inflammation in cancer-related anemia. 2005;106(1):362–7. doi: 10.1182/blood-2005-01-0160.

  16. Сахин В.Т., Маджанова Е.Р., Крюков Е.В. и др. Патогенетические особенности анемии у больных с солидными опухолями. Клиническая онкогематология. 2017;10(4):514–8. doi: 10.21320/2500-2139-2017-10-4-514-518.

    [Sakhin VT, Madzhanova ER, Kryukov EV, et al. Pathogenetic Characteristics of Anemia in Patients with Solid Tumors. Clinical oncohematology. 2017;10(4):514–8. doi: 10.21320/2500-2139-2017-10-4-514-518. (In Russ)]

  17. Park S, Jung CW, Kim K, et al. Iron deficient erythropoiesis might play key role in development of anemia in cancer patients. Oncotarget. 2015;6(40):42803–12. doi: 10.18632/oncotarget.5658.

  18. Speeckaert MM, Speeckaert R, Delanghe JR. Biological and clinical aspects of soluble transferrin receptor. Crit Rev Clin Lab Sci. 2010;47(5–6):213–28. doi: 10.3109/10408363.2010.550461.

  19. Moldawer LL, Marano MA, Wei H, et al. Cachectin/tumor necrosis factor-alpha alters red blood cell kinetics and induces anemia in vivo. FASEB J. 1989;3(5):1637–43.

  20. Raj DSC. Role of interleukin-6 in the anemia of chronic disease. Sem Arthritis Rheum. 2009;38(5):382–8. doi: 10.1016/j.semarthrit.2008.01.006.

  21. Wrighting DM, Andrews NC. Interleukin-6 induces hepcidin expression through STAT3. Blood. 2006;108(9):3204–9. doi: 10.1182/blood-2006-06-027631.

  22. Huang P, Wang J, Lin X, et al. Effects of IL-10 on iron metabolism in LPS-induced inflammatory mice via modulating hepcidin expression. Eur Rev MedPharmacol Sci. 2017;21(15):3469–75.

  23. Shanmugam NKN, Ellenbogen S, Trebicka E, et al. Tumor necrosis factor α inhibits expression of the iron regulating hormone hepcidin in murine models of innate colitis. PLoS One. 2012;7(5):e38136. doi: 10.1371/journal.pone.0038136.

  24. De Lurdes Cabrita AA, Pinho A, Malho A, et al. Risk factors for high erythropoiesis stimulating agent resistance index in pre-dialysis chronic kidney disease patients, stages 4 and 5. Int Urol Nephrol. 2011;43(3):835–40. doi: 10.1007/s11255-010-9805-9.

  25. Nazemian F, Karimi G, Moatamedi M, et al. Effect of silymarin administration on TNFalpha serum concentration in peritoneal dialysis patients. Phytother Res. 2010;24(11):1654–7. doi: 10.1002/ptr.3175.

Pure Red Cell Aplasia with M-Gradient: A Literature Review and Clinical Experience

AV Pivnik1,2, AA Petrenko1, SV Kozhurin3, SA Mar’ina3

1 RUDN University, 6 Miklukho-Maklaya str., Moscow, Russian Federation, 117198

2 AS Loginov Moscow Clinical Scientific Center, 89 Entuziastov sh., Moscow, Russian Federation, 111123

3 National Medical Hematology Research Center, 4a Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Prof. Aleksandr Vasil’evich Pivnik, MD, PhD, 89 Entuziastov sh., Moscow, Russian Federation, 111123; Tel.: 8(495)304-30-39; e-mail: pivnikav@gmail.com

For citation: Pivnik AV, Petrenko AA, Kozhurin SV, Mar’ina SA. Pure Red Cell Aplasia with M-Gradient: A Literature Review and Clinical Experience. Clinical oncohematology. 2018;11(3):273–80.

DOI: 10.21320/2500-2139-2018-11-3-273-280


ABSTRACT

Background. Pure red cell aplasia (PRCA) is a rare syndrome characterized by a decrease of erythroid progenitor cell count in the bone marrow. M-gradient with both a light and a heavy chain types in PRCA patients is a rare phenomenon which is considered to be a specific form of the disease.

Aim. To review a clinical presentation, diagnostic capabilities, and treatment outcomes of PRCA with M-gradient.

Materials & Methods. The analysis included 10 patients. The most effective empirically established treatment program was 200–400 g of cyclophosphamide 2–3 times a week to a total dose of 6–10 g and loading courses of 100–120 mg of oral and 180–240 mg of intravenous prednisone daily within 5 days. On the 6th day prednisone injections were discontinued, and from the 7th day the oral dose of prednisone was gradually reduced to permanent discontinuation in 2–3 days. This treatment course was repeated 1–3 times at intervals of a week. Targeted enzyme immunoassay of M-gradient was performed in 4 patients in order to determine whether M-gradient is the sum of two antibody types, i.e. erythrokaryocyte antibodies and secondary anti-idiotype antibodies against primary antibodies.

Results. The total of 6 out of 10 PRCA patients reached complete remission within the period from 9 months to 22 years of follow-up, in 4 patients no remission was achieved. M-gradient contained IgG (n = 9) and IgA (n = 1) oligoclones. In typing it consisted of IgGλ (n = 4), IgGκ (n = 5) and IgAκ (n = 1). M-gradient enzyme immunoassay showed no primary and secondary anti-idiotype antibodies.

Conclusion. The obtained results allow to regard gammopathy in PRCA as an effect of oligoclonal hyper-immunoglobulin without any pathogenetic connection between M-gradient and PRCA.

Keywords: partial red cell aplasia of the bone marrow, PRCA, anemia, M-gradient, monoclonal gammopathy.

Received: February 5, 2018

Accepted: May 11, 2018

Read in PDF 


REFERENCES

  1. Japson JH, Lovenstein L. Panhypoplasia in the bone marrow. 1. Demonstration of a plasma factor with anti-erythropoietin-like activity. Can Med Assoc J. 1968;99(3):99–101.
  2. Гериатрическая гематология. Заболевания системы крови в старших возрастных группах. Под ред. Л.Д. Гриншпун, А.В. Пивника. М.: Медиум, 2012. Т. 2. С. 438–41.[Grinshpun LD, Pivnik AV, eds. Geriatricheskaya gematologiya. Zabolevaniya sistemy krovi v starshikh vozrastnykh gruppakh. (Geriatric hematology. Blood disorders in elderly patients.) Moscow: Medium Publ.; 2012, vol. 2. pp. 438–41. (In Russ)]
  3. Charles RJ, Sabo KM, Kidd PG, Abkowitz JL. The pathophysiology of pure red cell aplasia: implications for therapy. Blood. 1996;87(11):4831–8.
  4. Lacy MQ, Kurtin PJ, Tefferi A. Pure red cell aplasia: association with large granular lymphocyte leukemia and the prognostic value of cytogenetic abnormalities. Blood. 1996;87(7):3000–6.
  5. Руководство по гематологии. Под ред. А.И. Воробьева. 3-е изд. перераб. и доп. М.: Ньюдиамед, 2005. Т. 3. С. 274–85.[Vorob’ev AI, ed. Rukovodstvo po gematologii. (Guidelines on hematology.) 3rd revised edition. Moscow: Newdiamed Publ.; 2005, vol. 3. pp. 274–85. (In Russ)]
  6. Идельсон Л.И., Тер-Григоров B.C., Пивник A.B., Эткин А.Ф. Клинико-иммунологические особенности парциальной красноклеточной аплазии сочетанной с моноклональной гаммапатией. Терапевтический архив. 1983;8:75–9.[Idel’son LI, Ter-Grigorov VC, Pivnik AV, Etkin AF. Clinical and immunological profile of pure red cell aplasia together with monoclonal gammopathy. Terapevticheskii arkhiv. 1983;8:75–9. (In Russ)]
  7. Пивник А.В. Парциальная красноклеточная аплазия костного мозга: клиника, вопросы патогенеза, диагностика, лечение: Дис. … канд. мед. наук. М., 1978.[Pivnik AV. Partsial’naya krasnokletochnaya aplaziya kostnogo mozga: klinika, voprosy patogeneza, diagnostika, lechenie. (Pure red cell aplasia in bone marrow: clinical presentation, pathogenesis, diagnostics, treatment.) [dissertation] Moscow; 1978. (In Russ)]
  8. Андреева Н.Е., Чернохвостова Е.В. Иммуноглобулинопатии. М.: Медицина, 1985. 240 c.[Andreeva NE, Chernokhvostova EV. Immunoglobulinopatii. (Immunoglobulinopathy.) Moscow: Meditsina Publ.; 1985. 240 p. (In Russ)]
  9. Эткин А.Ф., Пивник А.В., Мамиляева З.Х. Олигоклональная парапротеинемия при парциальной аплазии костного мозга. Гематология и трансфузиология. 1987;4:31–4.[Etkin AF, Pivnik AV, Mamilyaeva ZKh. Oligoclonal paraproteinemia in partial red cell aplasia in the bone marrow. Gematologiya i transfuziologiya. 1987;4:31–4. (In Russ)]
  10. Kobayashi T, Hanada T, Sato Y, et al. A case of pure red cell aplasia with monoclonal gammopathy: immune-mediated inhibition of erythropoiesis. Rinsho Ketsueki. 1987;28(11):2029–33.
  11. Balducci L, Hardy C, Dreiling B, et al. Pure red blood cell aplasia associated with paraproteinemia: in vitro studies of erythropoiesis. Haematologia (Budap). 1984;17(3):353–7.
  12. Gu H, Lee WI, Jeon Y, et al. Pure Red Cell Aplasia Associated with Monoclonal Gammopathy of Undetermined Significance and Literature Review. Clin Lab. 2017;63(2):373–8. doi: 10.7754/Clin.Lab.2016.160730.
  13. Murate T, Ohashi H, Ichikawa A, et al. A case of pure red cell aplasia with monoclonal gammopathy. Nihon Naika Gakkai Zasshi. 1992;81(8):1263–5.
  14. Shimmyozu K, Tara M, Okadome T, et al. Pure red cell aplasia with monoclonal gammopathy and von Willebrand disease. Rinsho Ketsueki. 1990;31(9):1468–73.
  15. Korde N, Zhang Y, Loeliger K, et al. Monoclonal gammopathy-associated pure red cell aplasia. Br J Haematol. 2016;173(6):876–83. doi: 10.1111/bjh.14012.
  16. Идельсон Л.И., Койфман М.М., Горина Л.Г., Оловников А.М. Применение агрегат-гемагглютинационной пробы и других методов для диагностики аутоиммунной гемолитической анемии с отрицательной прямой пробой Кумбса. Проблемы гематологии. 1975;6:91–9.[Idel’son LI, Koifman MM, Gorina LG, Olovnikov AM. Application of an aggregate-hemagglutination test and other assays for the diagnosis of autoimmune hemolytic anaemia with a negative direct antiglobulin test. Problemy gematologii. 1975;6:91–9. (In Russ)]

Correction of Anemia and Evaluation of Efficacy of Red Blood Cell Transfusion in Patients with Oncohematological Diseases

NA Romanenko1, AV Chechetkin1, LYu Zhiguleva1, GV Grishina1, SV Bondarchuk2, SS Bessmel’tsev1

1 Russian Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

2 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

For correspondence: Nikolai Aleksandrovich Romanenko, MD, PhD, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: 8(812)717-58-57; Fax: 8(812)717-67-80; е-mail: rom-nik@yandex.ru

For citation: Romanenko NA, Chechetkin AV, Zhiguleva LYu, et al. Correction of Anemia and Evaluation of Efficacy of Red Blood Cell Transfusion in Patients with Oncohematological Diseases. Clinical oncohematology. 2018;11(3):265–72.

DOI: 10.21320/2500-2139-2018-11-3-265-272


ABSTRACT

Aim. To study the quality of life (QL) of patients with oncohematological diseases and anemia with respect to hemoglobin level and to evaluate the efficacy of red blood cell transfusion (RBCT).

Materials & Methods. QL of patients (n = 326) was studied using FACT-An questionnaire. RBCT efficacy was evaluated in two groups. The first group included patients (n = 28; 13 men and 15 women) with oncohematological diseases and chronic anemia aged 23–80 (median 65) years, the second (control) group included patients (n = 12; 11 men and 1 woman) after severe blood loss after injury (acute anemia) aged 25–43 (median 36) years. The baseline levels of hemoglobin (Hb) and hematocrit (Ht) were < 80 g/L and < 25 % in all patients, respectively. The target levels of Hb and Ht were > 80 g/L and > 25 %, respectively.

Results. The association between the severity of anemia and QL was shown. The lowest QL was observed in patients with grade III–IV anemia (Hb < 80 g/L). Each patient in both groups received 1–8 units of blood cells (median 2) during the hospital stay. After RBCT the levels of Hb and Ht increased from 64.1 ± 2.7 g/L to 90.2 ± 1.7 g/L and from 20.1 ± 0.8 % to 28.9 ± 0.7 %, respectively. The levels of Hb and Ht in the second (control) group increased from 65.9 ± 3.0 g/L to 88.3 ± 3.2 g/L and from 19.6 ± 0.9 % to 26.7 ± 1.4 %, respectively. Venous blood oxygen saturation (SvO2) increased in the first group from 42.0 ± 3.3 % to 57.6 ± 4.1 %, and in the second group from 51.3 ± 1.9 % to 69.0 ± 1.3 %. However, after RBCT the SvO2 level reached > 60 % only in 67.9 % of patients in the first group and in all the patients (100 %) in the second group. In 32.1 % of patients with various forms of hematologic cancer and chronic anemia tissue hypoxia was still observed after RBCT despite increased Hb > 80 g/L and Ht > 25 %. Therefore, it was proposed to raise the target Hb and Ht threshold levels for patients with low SvO2.

Conclusion. The effect of the severity of anemia on QL was demonstrated. The patients with Hb < 80 g/L were shown to have low quality of life. SvO2 determination in anemia patients proved to be of great importance for RBCT efficacy evaluation. In patients with low SvO2 (< 60 %) RBCT should be continued until the target levels of Hb 100 g/L and Ht 33 % are reached.

Keywords: anemia, chronic anemia, red blood cell transfusions, hemoglobin concentration, hematocrit, venous blood oxygen saturation, quality of life, FACT-An questionnaire.

Received: March 10, 2018

Accepted: May 30, 2018

Read in PDF 


REFERENCES

  1. Сахин В.Т., Маджанова Е.Р., Крюков Е.В. и др. Патогенетические особенности анемии у больных с солидными опухолями. Клиническая онкогематология. 2017;10(4):514–8. doi: 10.21320/2500-2139-2017-10-4-514-518.[Sakhin VT, Madzhanova ER, Kryukov EV, et al. Pathogenetic Characteristics of Anemia in Patients with Solid Tumors. Clinical oncohematology. 2017;10(4):514–8. doi: 10.21320/2500-2139-2017-10-4-514-518. (In Russ)]
  2. Santos FPS, Alvarado Y, Kantarjian H, et al. Long-term prognostic impact of the use of erythropoietic-stimulating agents in patients with chronic myeloid leukemia in chronic phase treated with imatinib. Cancer. 2011;117(5):982–91. doi: 10.1002/cncr.25533.
  3. Steurer M, Wagner H, Gastel G. Prevalence and management of anaemia in haematologic cancer patients receiving cyclic nonplatinum chemotherapy: results of a prospective national chart survey. Wien Klin Wochenschr. 2004;116(11–12):367–72. doi: 1007/bf03040915.
  4. Романенко Н.А. Патогенез и терапия анемии препаратами рекомбинантного эритропоэтина у онкогематологических больных (обзор современной литературы). Онкогематология. 2012;7(3):22–9. doi: 10.17650/1818-8346-2012-7-3-22-29.[Romanenko NA. Pathogenesis and therapy of anemia in oncohematology patients with recombinant erythropoietin agent (literature review). Onkogematologiya. 2012;7(3):22–9. doi: 10.17650/1818-8346-2012-7-3-22-29. (In Russ)]
  5. Truong PT, Parhar T, Hart J, et al. Population-based analysis of the frequency of anemia and its management before and during chemotherapy in patients with malignant lymphoma. Am J Clin. Oncol. 2010;33(5):465–8. doi: 10.1097/COC.0b013e3181b4b147.
  6. Leitch HA, Vickars LM. Supportive care and chelation therapy in MDS: are we saving lives or just lowering iron? Hematology. 2009;2009(1):664–72. doi: 10.1182/asheducation-2009.1.664.
  7. Passamonti F, Rumi E, Arcaini L, et al. Blast phase of essential thrombocythemia: A single center study. Am J Hematol. 2009;84(10):641–4. doi: 10.1002/ajh.21496.
  8. Tefferi A, Lasho TL, Jimma T, et al. One thousand patients with primary myelofibrosis: the Mayo clinic experience. Mayo Clin Proc. 2012;87(1):25–33. doi: 10.1016/j.mayocp.2011.11.001.
  9. Quintas-Cardama A, De Souza Santos FP, Kantarjian H, et al. Dynamics and management of cytopenias associated with dasatinib therapy in patients with chronic myeloid leukemia in chronic phase after imatinib failure. Cancer. 2009;115(17):3935–43. doi: 10.1002/cncr.24432.
  10. Romanenko N, Abdulkadyrov K, Gritsaev S, Bessmeltsev S. Study of effectiveness recombinant human erythropoietin in chronic myeloid leukemia patients with anemia induced imatinib therapy. Haematologica. 2011;96(Suppl. 2):S
  11. Бессмельцев С.С., Романенко Н.А., Потихонова Н.А. и др. Злокачественные лимфопролиферативные заболевания с анемией: изменение качества жизни пациентов на фоне переливаний донорских эритроцитов и применения препаратов рекомбинантного эритропоэтина. Клиническая онкогематология. 2015;8(4):368–78. doi: 10.21320/2500-2139-2015-8-4-368-378.[Bessmel’tsev SS, Romanenko NA, Potikhonova NA, et al. Malignant Lymphoproliferative Disorders with Anemia: Changes of Quality of Life in Patients Treated with Donor Red Blood Cell Transfusions and Recombinant Erythropoietin. Clinical oncohematology. 2015;8(4):368–78. doi: 10.21320/2500-2139-2015-8-4-368-378. (In Russ)]
  12. Samuelsson J. Long-standing resolution of anemia in symptomatic low-grade non-Hodgkin’s lymphoma patients treated with recombinant human erythropoietin as sole therapy. Med Oncol. 2002;19(1):69–72. doi: 1385/MO:19:1:69.
  13. Романенко Н.А., Бессмельцев С.С., Потихонова Н.А. и др. Качество жизни больных лимфопролиферативными заболеваниями с анемией на фоне трансфузий эритроцитов и эритропоэзстимулирующих препаратов. Биомедицинский журнал Medline.ru. 2014;15(56):703–17.[Romanenko NA, Bessmel’tsev SS, Potikhonova NA, et al. Quality of life of anemia patients with lymphoproliferative disorders treated with red blood cell transfusions and erythropoiesis stimulating drugs. Biomeditsinskii zhurnal Medline.ru. 2014;15(56):703–17. (In Russ)]
  14. Romanenko N, Bessmeltsev S, Romanenko A, et al. Quality of life in anemic patients with hematological malignancies. Haematologica. 2017;102(Suppl 1):843.
  15. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: МК, 2016. 504 с.[Bessmel’tsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: guidelines for doctors.) Moscow: MK Publ.; 2016. 504 p. (In Russ)]
  16. Романенко Н.А., Бессмельцев С.С., Чечеткин А.В. Коррекция иммунного статуса пациентов иммуноглобулином человека для внутривенного введения. Казанский медицинский журнал. 2017;98(5):775–83.[Romanenko NA, Bessmel’tsev SS, Chechetkin AV. Correction of patients’ immune status with human intravenous immunoglobulin. Kazanskii meditsinskii zhurnal. 2017;98(5):775–83. (In Russ)]
  17. Романенко Н.А., Головченко Р.А., Бессмельцев С.С. и др. Эффективность трансфузий донорских эритроцитов у больных гемобластозами с анемией. Трансфузиология. 2015;16(2):29–42.[Romanenko NA, Golovchenko RA, Bessmel’tsev SS, et al. Efficacy of donor red blood cell transfusions in patients with blood cancer and anemia. Transfuziologiya. 2015;16(2):29–42. (In Russ)]
  18. Romanenko N, Potikhonova N, Tiranova S, et al. Dynamics of Quality of Life in anemic patients with lymphoproliferative disorders treated with red blood cell transfusions and erythropoiesis-stimulating agents. 2016;101(Suppl 1):S40.
  19. Приказ МЗ РФ № 363 от 25.11.2002 г. «Об утверждении Инструкции по применению компонентов крови»). М., 2002.[Decree No. 363 of RF MH dated November 25, 2002. Approval of the Instruction on use of blood components. Mоscow; 2002. (In Russ)]
  20. Provan D, Baglin T, Dokal I, de Vos J. Oxford Handbook of Clinical Haematology, 4th edition. Oxford University Press; 2015. 805 p. doi: 10.1093/med/9780199683307.001.0001.
  21. Бессмельцев С.С., Романенко Н.А. Анемия при опухолевых заболеваниях системы крови: руководство для врачей. М.: СИМК, 2017. 228 с.[Bessmel’tsev SS, Romanenko NA. Anemiya pri opukholevykh zabolevaniyakh sistemy krovi: rukovodstvo dlya vrachei. (Anemia in patients with blood cancer: guidelines for doctors.) Moscow: SIMK Publ.; 2017. 228 p. (In Russ)]
  22. Трансфузиология: Клиническое руководство. Под ред. М.Ф. Заривчацкого. Пермь: ГБОУ ВПО ПГМА им. акад. Е.А. Вагнера Минздрава России, 2014. 900 с.[Zarivchatskii MF, ed. Transfuziologiya: Klinicheskoe rukovodstvo. (Transfusiology: clinical guidelines.) Perm: E.A. Vagner Medical University Publ.; 2014. 900 p. (In Russ)]
  23. Приказ МЗ РФ № 183н от 2.04.2013 г. «Об утверждении правил клинического использования донорской крови и (или) ее компонентов». М., 2013.[Decree No. 183н of RF MH dated April 2, 2013. Approval of the Instruction on clinical use of donated blood and/or its components. Mоscow; 2013. (In Russ)]
  24. Чечеткин А.В., Данильченко В.В., Шайдаков Е.В. и др. Трансфузионная терапия послеоперационной анемии у больных при плановых хирургических вмешательствах в военных лечебных учреждениях. Методические рекомендации. СПб., 2006. 28 c.[Chechetkin AV, Danil’chenko VV, Shaidakov EV, et al. Transfuzionnaya terapiya posleoperatsionnoi anemii u bol’nykh pri planovykh khirurgicheskikh vmeshatel’stvakh v voennykh lechebnykh uchrezhdeniyakh. Metodicheskie rekomendatsii. (Transfusion therapy of post-surgery anemia in scheduled surgeries in military medical institutions. Guidelines.) Saint Petersburg; 2006. 28 p. (In Russ)]
  25. Романенко Н.А., Грицаев С.В., Бессмельцев С.С., Абдулкадыров К.М. Эффективность эритропоэзстимулирующих препаратов при анемии у больных миелодиспластическим синдромом. Гематология и трансфузиология. 2013;58(4):18–21.[Romanenko NA, Gritsaev SV, Bessmel’tsev SS, Abdulkadyrov KM. Efficacy of erythropoiesis stimulating drugs in anemia patients with myelodysplastic syndrome. Gematologiya i transfuziologiya. 2013;58(4):18–21. (In Russ)]

Pathogenetic Characteristics of Anemia in Patients with Solid Tumors

VT Sakhin1, ER Madzhanova1, EV Kryukov3, AV Sotnikov2, AV Gordienko2, OA Rukavitsyn3

11586 Military Clinical Hospital, 4 Mashtakova str., Podolsk, Russian Federation, 142110

2SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

3NN Burdenko Main Military Clinical Hospital, 3 Gospitalnaya sq., Moscow, Russian Federation, 105229

For correspondence: Valery Timofeevich Sakhin, PhD, 4 Mashtakova str., Podolsk, Russia, 142110; Tel.: +7(916)314-31-11; e-mail: SahinVT@yandex.ru

For citation: Sakhin VT, Madzhanova ER, Kryukov EV, et al. Pathogenetic Characteristics of Anemia in Patients with Solid Tumors. Clinical oncohematology. 2017;10(4):514–8 (In Russ).

DOI: 10.21320/2500-2139-2017-10-4-514-518


ABSTRACT

Aim. To study the impact iron metabolism disturbances and cytokine levels on the development of anemia in patients with solid tumors.

Materials & Methods. The research included 42 patients with malignant neoplasms, including 24 patients with anemia (19 men and 5 women, median age 67.7 ± 10 years) and 18 patients without anemia (15 men, 3 women, median age 65.7 ± 14 years). Anemia was diagnosed according to the WHO criteria (in men: erythrocytes < 4.0 × 1012/L, hemoglobin < 130 g/L, hematocrit < 39 %; in women: erythrocytes < 3.8 × 1012/L, hemoglobin < 120 g/L, hematocrit < 36 %).

Results. A comparative analysis of iron metabolism in patients with and without anemia was performed. The lower values of serum iron and transferrin saturation in patients with anemia were shown (< 0.05). The total iron-binding capacity, the levels of ferritin, transferrin, C-reactive protein, indirect bilirubin were similar between groups (> 0,05). Higher levels of interleukins 6 and 10 (IL-6 and IL-10) were observed in patients with anemia (< 0.05). For IL-6, correlations were observed with levels of erythrocytes (r = –0,58), hemoglobin (r = –0,57), hematocrit (r = –0,52), and leukocytes (r = 0,42). The levels of IL-10 slightly correlated with the levels of erythrocytes, leukocytes, platelets, MCV, and MCH (r < 0.3). For IL-10, correlations were established with levels of MCHC (r = –0,71), hemoglobin (r = –0,64) and hematocrit (r = –0,32). Correlations between the levels IL-6, IL-6 and hemoglobin, erythrocytes and several color indices may indicate their influence on the development of anemia in patients with malignant neoplasms.

Conclusion. A functional iron deficiency in patients with anemia was found. Several causes of anemia development and significant role of interleukins in anemia pathogenesis were also discovered.

Keywords: cancer, anemia, iron metabolism, interleukin-6, interleukin-10.

Received: March 21, 2017

Accepted: June 22, 2017

Read in PDF


REFERENCES

  1. Maccio A, Madeddu C, Gramignano G, et al. The role of inflammation, iron, and nutritional status in cancer-related anemia: results of a large, prospective, observational study. Haematologica. 2015;100(1):124–32. doi: 10.3324/haematol.2014.112813.
  2. Ludwig H, Van Belle S, Barrett-Lee P, et al. The European Cancer Anaemia Survey (ECAS): A large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur J Cancer. 2004;40(15):2293–306. doi: 10.1016/j.ejca.2004.06.019.
  3. Steinmetz T, Totzke U, Schweigert M, et al. A prospective observational study of anaemia management in cancer patients–results from the German Cancer Anaemia Registry. Eur J Cancer Care. 2011;20(4):493–502. doi: 10.1111/j.1365-2354.2010.01230.x.
  4. Waters JS, O’Brien MER, Ashley S. Management of anemia in patients receiving chemotherapy. J Clin Oncol. 2002;20(2):601–3. doi: 10.1200/JCO.2002.20.2.601.
  5. Grotto HZ. Anaemia of cancer: an overview of mechanisms involved in its pathogenesis. Med Oncol. 2008;25(1):12–21. doi: 10.1007/s12032-007-9000-8.
  6. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. С. 143–9.[Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. pp. 143–9 (In Russ)]
  7. Ludwig H, Muldur E, Endler G, et al. Prevalence of iron deficiency across different tumors and its association with poor performance status, disease status and anemia. Ann Oncol. 2013;24(7):1886–92. doi: 10.1093/annonc/mdt118.
  8. de Castro J, Gascоn P, Casas A, et al. Iron deficiency in patients with solid tumours: prevalence and management in clinical practice. Clin Transl Oncol. 2014;16(9):823–8. doi: 10.1007/s12094-013-1155-5.
  9. Beguin Y. Prediction of response and other improvements on the limitations of recombinant human erythropoietin therapy in anemic cancer patients. Haematologica. 2002;87(11):1209–21.
  10. Steinmetz HT, Tsamaloukas A, Schmitz S, et al. A new concept for the differential diagnosis and therapy of anaemia in cancer patients. Support Care Cancer. 2010;19(2):261–9. doi: 10.1007/s00520-010-0812-2.
  11. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011–23. doi: 10.1056/nejmra041809.
  12. Maccio A, Madeddu C, Massa D, et al. Hemoglobin levels correlate with interleukin-6 levels in patients with advanced untreated epithelial ovarian cancer: role of inflammation in cancer-related anemia. Blood. 2005;106(1):362–7. doi: 10.1182/blood-2005-01-0160.
  13. Falkensammer CE, Thurnher M, Leonhartsberger N, Ramoner R. C-reactive protein is a strong predictor for anaemia in renal cell carcinoma: role of IL-6 in overall survival. BJU Int. 2011;107(12):1893–8. doi: 10.1111/j.1464-410x.2010.09817.x.

Anemias in Oncology: Potential of Maintenance Therapy

AV Snegovoi, VB Larionova, LV Manzyuk, IB Kononenko

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Anton Vladimirovich Snegovoi, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-41-09; e-mail: anvs2012@gmail.com

For citation: Snegovoi AV, Larionova VB, Manzyuk LV, Kononenko IB. Anemias in Oncology: Potential of Maintenance Therapy. Clinical oncohematology. 2016;9(3):326-35 (In Russ).

DOI: 10.21320/2500-2139-2016-9-3-326-35


ABSTRACT

Development of anemia during chemotherapy or chemoradiotherapy of malignancies is a serious adverse event negatively affecting the quality of life and effectiveness of the treatment. In this regard, the leading consensus committees of NCCN, ESMO, ASCO, and RUSSCO have prepared and are regularly updating recommendations for diagnosing and treatment of anemia in cancer patients. The article presents recent data on the pathogenesis and methods of treatment of anemia in cancer patients, including administration of erythropoiesis stimulating agents: recombinant erythropoietins, intravenous iron products, vitamins, and red blood cell transfusions.


Keywords: cancer, anemia, erythropoietins, intravenous iron products, red blood cell transfusions.

Received: February 29, 2016

Accepted: March 31, 2016

Read in PDF(RUS)pdficon


REFERENCES

  1. Knight K, Wade S, Balducci L. Prevalence and outcome of anemia in cancer: a systematic review of the literature. Am J Med. 2004;116(7);11–26. doi: 10.1016/j.amjmed.2003.12.008.
  2. Schrijvers D, De Samblanx H, Roila F. Erythropoiesis-stimulating agents in the treatment of anaemia in cancer patients: ESMO Clinical Practice Guidelines for use. Ann Oncol. 2010;21(Suppl 5):v244–v247. doi: 10.1093/annonc/mdq202.
  3. Ludwig H, Van Belle S, Barrett-Lee P, et al. The European Cancer Anaemia Survey (ECAS): A large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur J Cancer. 2004;40(15):2293–306. doi: 10.1016/j.ejca.2004.06.019.
  4. Groopman J, Itri L. Chemotherapy-induced anemia in adults: incidence and treatment. J Nat Cancer Inst. 1999;91(19):1616–34. doi: 10.1093/jnci/91.19.1616.
  5. Food and Drug Administration. Jevtana (cabazitaxel) Injection Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/201023lbl.pdf. (accessed 12.11.2015).
  6. Food and Drug Administration. Taxotere (docetaxel) Injection Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020449s059lbl.pdf (accessed 12.11.2015).
  7. Food and Drug Administration. Xtandi (enzalutamide) Capsules Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203415lbl.pdf. (accessed 12.11.2015).
  8. Cella D. Quality of life and clinical decisions in chemotherapy-induced anemia. Oncology (Williston Park). 2006;20(8 Suppl 6):25–8.
  9. Caro JJ, Salas M, Ward A, et al. Anemia as an independent prognostic factor for survival in patients with cancer: a systemic, quantitative review. Cancer. 2001;91(12):2214–21. doi: 10.1002/1097-0142(20010615)91:12<2214::aid-cncr1251>3.0.co;2-p.
  10. Glaspy J. Update on Safety of ESAs in Cancer-Induced Anemia. J Natl Compr Cancer Network. 2012;10(5):659–66.
  11. Nowrousian M, Smyth JF, et al, eds. rhErythropoietin in cancer supportive treatment. New York: Marcel Dekker Inc; 1996. pp. 13–34.
  12. Павлов А.Д., Морщакова Е.Ф., Румянцев А.Г. Эритропоэз, эритропоэтин, железо. Молекулярные и клинические аспекты. М.: ГЭОТАР-Медиа, 2011. 304 с.
    [Pavlov AD, Morshchakova EF, Rumyantsev AG. Eritropoez, eritropoetin, zhelezo. Molekulyarnye i klinicheskie aspekty. (Erythropoiesis, erythropoietin, and iron. Molecular and clinical aspects.) Moscow: GEOTAR-Media Publ.; 2011. 304 p. (In Russ)]
  13. Grotto HZ. Anaemia of cancer: an overview of mechanisms involved in its pathogenesis. Med Oncol. 2008;25(1):12–21. doi: 10.1007/s12032-007-9000-8.
  14. Aapro M, Osterborg A, Gascon P, et al. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol. 2012;23(8):1954–62. doi: 10.1093/annonc/mds112.
  15. Khorana AA, Francis CW, Blumberg N, et al. Blood transfusions, thrombosis and mortality in hospitalized cancer patients. Arch Int Med. 2008;168(21):2377–81. doi: 10.1001/archinte.168.21.2377.
  16. Amato A, Pescatori M. Perioperative blood transfusions for the recurrence of colorectal cancer. Cochrane database syst.rev. 2006;25(1):CD005033. doi: 10.1002/14651858.cd005033.pub2.
  17. Hoff CM, Lassen P, Eriksen JG, et al. Does transfusion improve the outcome for HNSCC patients treated with radiotherapy? Results from the randomized DAHANCA 5 and 7 trials. Acta Oncologica. 2011;50(7):1006–14. doi: 10.3109/0284186X.2011.592650.
  18. Egrie JC, Browne JK. Development and characterization of novel erythropoiesis stimulating protein (NESP). Br J Cancer. 2001;84(Suppl 1):3–10. doi: 10.1054/bjoc.2001.1746.
  19. Bohlius J, Wilson J, Seidenfeld J, et al. Recombinant human erythropoietins and cancer patients: updated meta-analysis of 57 studies including 9353 patients. J Natl Cancer Inst. 2006;98(10):708–14. doi: 10.1093/jnci/djj189.
  20. Tonelli M, Hemmelgarn B, Reiman T, et al. Benefits and harms of erythropoiesis-stimulating agents for anemia related to cancer: a meta-analysis. CMAJ. 2009;180(11):E62–E71. doi: 10.1503/cmaj.090470.
  21. Bennett C, Silver S, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA. 2008;299(8):914–24. doi: 10.1001/jama.299.8.914.
  22. Ludwig H, Crawford J, Osterborg A, et al. Pooled analysis of individual patient-level data from all randomized, double-blind, placebo-controlled trials of darbepoetin alfa in the treatment of patients with chemotherapy-induced anemia. J Clin Oncol. 2009;27(17):2838–47. doi: 10.1200/jco.2008.19.1130.
  23. Glaspy J, Crawford J, Vansteenkiste J, et al. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. Br J Cancer. 2010;102(2):301–15. doi: 10.1038/sj.bjc.6605498.
  24. Oncologic Drug Advisory Committee (ODAC) Meeting Information Package. Darbepoetin alfa (BLA #103951) and Epoetin alfa (BLA #103234). [Internet] Available from: http://www.scribd.com/doc/1117102/US-Food-and-Drug-Administration-20074301b20101Amgen. (accessed 20.02.2016).
  25. Glaspy J, Osterborg A, Ludwig H, et al. Evaluation of the association between (Hb) events and safety outcomes in cancer patients with chemotherapy induced anemia: an integrated analysis of patient-level data from 6 randomized, placebo-controlled trials of darbepoetin. Eur J Cancer. 2007;5(4):147–8. doi: 10.1016/s1359-6349(07)70639-0.
  26. Снеговой А.В., Aapro M., Давиденко И.С. и др. Практические рекомендации по лечению анемии у онкологических больных. Злокачественные опухоли. 2015;4:316–26. doi: 10.18027/2224-5057-2015-4s-316-326. [Snegovoi AV, Aapro M, Davidenko IS, et al. Practical recommendations for management of anemia in cancer patients. Zlokachestvennye opukholi. 2015;4:316–26. doi: 10.18027/2224-5057-2015-4s-316-326. (In Russ)]
  27. Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. The Lancet. 2003;362(9392):1255–60. doi: 10.1016/s0140-6736(03)14567-9.
  28. Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol. 2005;23(25):5960–72. doi: 10.1200/jco.2005.06.150.
  29. Delarue R, Haioun C, Coiffier B, et al. Survival effect of darbepoetin alfa in patients with diffuse large B-cell lymphoma (DLBCL) treated with immunochemotherapy: The LNH03-6B study. J Clin Oncol. 2011;29: Abstract 9048.
  30. Nitz U, Gluz O, Oberhoff C, et al. Adjuvant chemotherapy with or without darbepoetin alpha in node-positive breast cancer: survival and quality of life analysis from the prospective randomized WSG ARA plus trial. Cancer Res. 2012;71(24 Suppl):PD07-06. doi: 10.1158/0008-5472.sabcs11-pd07-06.
  31. National Institute for Health and Care Excellence. Health Technology Appraisal Programme. Equality impact assessment – Guidance development. MTA Erythropoiesis-stimulating agents (epoetin and darbepoetin) for treating anaemia in people having cancer treatment (including review of TA142). [Internet] Available from: https://www.nice.org.uk/guidance/ta323/documents/anaemia-cancertreatment-induced-erythropoiesisstimulating-agents-epoetin-and-darbepoetin-inc-rev-ta142-equality-impact-assessment-guidance-development2. (accessed 06.06.2016).
  32. Aapro M, Osterborg A, Gascon P, et al. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol. 2012;23(8):1954–62. doi: 10.1093/annonc/mds112.
  33. Beale AL, Penney MD, Allison MC. The prevalence of iron deficiency among patients presenting with colorectal cancer. Colorectal Dis. 2005;7(4):398–402. doi: 10.1111/j.1463-1318.2005.00789.x.
  34. Kuvibidila S, Gauthier T, Rayford W. Increased levels of serum transferrin receptor and serum transferrin receptor/log ferritin ratios in men with prostate cancer and the implications for body-iron stores. J Lab Clin Med. 2004;144(4):176–82. doi: 10.1016/j.lab.2004.03.017.
  35. Steinmetz HT, Tsamaloukas A, Schmitz S, et al. A new concept for the differential diagnosis and therapy of anaemia in cancer patients. Supp Care Cancer. 2010;19(2):261–9. doi: 10.1007/s00520-010-0812-2.
  36. Beguin Y, Lybaert W, Bosly A. A prospective observational study exploring the impact of iron status on response to darbepoetin alfa in patients with chemotherapy induced anemia. Blood. 2009;114(22): Abstract 2007.
  37. Ludwig H, Muldur E, Endler G, et al. High prevalence of iron deficiency across different tumors correlates with anemia, increases during cancer treatment and is associated with poor performance status. Haematologica. 2011;96: Abstract 982.
  38. Dangsuwan P, Manchana T. Blood transfusion reduction with intravenous iron in gynecologic cancer patients receiving chemotherapy. Gynecol Oncol. 2010;116(3):522–5. doi: 10.1016/j.ygyno.2009.12.004.
  39. Kim YT, Kim SW, Yoon BS, et al. Effect of intravenously administered iron sucrose on the prevention of anemia in the cervical cancer patients treated with concurrent chemoradiotherapy. Gynecol Oncol. 2007;105(1):199–204. doi: 10.1016/j.ygyno.2006.11.014.
  40. Steinmetz HT. The role of intravenous iron in the treatment of anemia in cancer patients. Ther Adv Hematol. 2012;3(3):177–91. doi: 10.1177/ 2040620712440071.
  41. Henry DH. Parenteral Iron Therapy in Cancer-Associated Anemia. Hematology. 2010;2010(1):351–6. doi: 10.1182/asheducation-2010.1.351.
  42. Auerbach M, Silberstein PT, Webb RT, et al. Darbepoetin alfa 300 or 500 mg once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. Am J Hematol. 2010;85(9):655–63. doi: 10.1002/ajh.21779.
  43. Bastit L, Vandebroek A, Altintas S, et al. Randomized, multicenter, controlled trial comparing the efficacy and safety of darbepoetin alpha administered every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. J Clin Oncol. 2008;26(10):1611–8. doi: 10.1200/jco.2006.10.4620.
  44. Henry DH, Dahl NV, Auerbach M, et al. Intravenous ferric gluconate significantly improves response to epoetin alfa versus oral iron or no iron in anemic patients with cancer receiving chemotherapy. The Oncologist. 2007;12(2):231–42. doi: 10.1634/theoncologist.12-2-231.
  45. Pedrazzoli P, Farris A, Del Prete S, et al. Randomized trial of intravenous iron supplementation in patients with chemotherapy-related anemia without iron deficiency treated with darbepoetin alpha. J Clin Oncol. 2008;26(10):1619–25. doi: 10.1200/jco.2007.12.2051.
  46. Steensma DP, Sloan JA, Dakhil SR, et al. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy-associated anemia. J Clin Oncol. 2011;29(1):97–105. doi: 10.1200/jco.2010.30.3644.
  47. Beguin Y, Maertens J, De Prijck B, et al. Darbepoetin-alfa and I.V. iron administration after autologous hematopoietic stem cell transplantation: a prospective randomized multicenter trial. Am J Hematol. 2013;88(12):990–6. doi: 10.1002/ajh.23552.
  48. Hedenus M, Birgegard G, Nasman P, et al. Addition of intravenous iron to epoetin beta increases hemoglobin response and decreases epoetin dose requirement in anemic patients with lymphoproliferative malignancies: a randomized multicenter study. Leukemia. 2007;21:627–32. doi: 10.1038/sj.leu.2404562.
  49. Gafter-Gvili A, Rozen-Zvi B, Vidal L, et al. Intravenous iron supplementation for the treatment of cancer-related anemia—systematic review and meta-analysis. Acta Oncologica. 2013;52(1):18–29. doi: 10.3109/0284186x.2012.702921.
  50. Petrelli F, Borgonovo K, Cabiddu M, et al. Addition of iron to erythropoiesis-stimulating agents in cancer patients: a meta-analysis of randomized trials. J Cancer Res Clin Oncol. 2012;138(2):179–87. doi: 10.1007/s00432-011-1072-3.
  51. Сельчук В.Ю., Чистяков С.С., Толокнов Б.О. и др. Железодефицитная анемия: современное состояние проблемы. Русский медицинский журнал. 2012;3(1):1–8.
    [Sel’chuk VYu, Chistyakov SS, Toloknov BO, et al. Iron-deficiency anemia: state-of-the-art of the problem. Russkii meditsinskii zhurnal. 2012;3(1):1–8. (In Russ)]

Malignant Lymphoproliferative Disorders with Anemia: Changes of Quality of Life in Patients Treated with Donor Red Blood Cell Transfusions and Recombinant Erythropoietin

S.S. Bessmeltsev1, N.A. Romanenko1, N.A. Potikhonova1, S.A. Tiranova1, M.N. Zenina1, A.E. Romanenko2, L.Yu. Zhiguleva1, K.M. Abdulkadyrov1

1 Russian Scientific Research Institute of Hematology and Transfusiology under the Federal Medico-Biological Agency, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

2 St. Olga Children Hospital, 2 Zemledelcheskaya str., Saint Petersburg, Russian Federation, 194156

For correspondence: Stanislav Semenovich Bessmel’tsev, DSci, Professor, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-58-57; e-mail: RNIIHT@mail.ru

For citation: Bessmel’tsev SS, Romanenko NA, Potikhonova NA, et al. Malignant Lymphoproliferative Disorders with Anemia: Changes of Quality of Life in Patients Treated with Donor Red Blood Cell Transfusions and Recombinant Erythropoietin. Clinical oncohematology. 2015;8(4):368–378 (In Russ).

DOI: 10.21320/2500-2139-2015-8-4-368-378


ABSTRACT

Aim. To evaluate the quality of life of anemic patients with malignant lymphoproliferative disorders after red blood cells transfusion and administration of human recombinant erythropoietin agents.

Methods. The efficacy was assessed based on evaluation of clinical data, lab test findings and quality of life (QoL) parameters using the FACT-An questionnaire. Red blood cells transfusions (RBCsT) were prescribed to anemic patients with malignant lymphoproliferative disorders (LPD) (n = 54) with initial Hb levels of 7.0 ± 0.16 g/dl. After red blood cells transfusion (Me = 3 Units) the Hb level increased up to 9.31 ± 0.12 g/dl. Human recombinant erythropoietin agents (EPO) were prescribed to LPD patients (n = 77) with initial Hb levels of 8.84 ± 0.14 g/dl.

Results. A positive response with a ³ 2.0 g/dl increase of Hb levels was observed in 52 (67.5 %) of 77 patients. The Hb level in responsive patients increased up to 12.31 ± 0.24 g/dl. Statistically significant QoL changes were observe according to «Physical well-being», «Emotional well-being», «Functional well-being», and «Anemia» scales. After EPO-therapy, significant changes were found according to «Physical well-being» and «Anemia» scales. A comparative analysis of QoL in both groups of patients demonstrated maximum changes according to the «Physical well-being» scale (from 12.9 ± 0.7 to 11.0 ± 0.8 points after RBCsT, < 0.001; from 11.6 ± 0.7 to 9.6 ± 0.7 points on EPO, < 0.02) and the «Anemia» scale (from 41.1 ± 2.0 to 34.2 ± 2.2 points after RBCsT, < 0.001; from 34.5 ± 1.7 to 30.1 ± 1.6 points on EPO; < 0.001).

Conclusion. Therefore, RBCsT and EPO therapy may significantly increase the Hb levels and improve QoL. However, the QoL of LPD patients on EPO proved to be better than that after RBCsT, because it had been possible to achieve normal and stable Hb levels.


Keywords: anemia, malignant lymphoproliferative disorders, erythropoietin, recombinant erythropoietin, donor’s red blood cells, red blood cell transfusions, quality of life, FACT-An questionnaire, «Anemia» scale.

Received: May 13, 2015

Accepted: October 12, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Бессмельцев С.С., Романенко Н.А., Абдулкадыров К.М. Современные подходы к лечению анемии у больных с онкогематологическими заболеваниями. Современная онкология. 2010;12(1):70–5.
    [Bessmel’tsev SS, Romanenko NA, Abdulkadyrov KM. Modern approaches to management of anemic patients with oncohematological disorders. Sovremennaya onkologiya. 2010;12(1):70–5. (In Russ)]
  2. Романенко Н.А., Абдулкадыров К.М. Патогенетическая коррекция анемии эритропоэзстимулирующими препаратами у больных лимфопролиферативными заболеваниями. Онкогематология. 2011;3:39–49.
    [Romanenko NA, Abdulkadyrov KM. Pathogenetic treatment of anemia using erythropoiesis-stimulating agents in patients with lymphoproliferative disorders. Onkogematologiya. 2011;3:39–49. (In Russ)]
  3. Steurer M, Wagner H, Gastel G. Prevalence and management of anaemia in haematologic cancer patients receiving cyclic nonplatinum chemotherapy: results of a prospective national chart survey. Wien Klin Wochenschr. 2004;116(11–12):367–72. doi: 10.1007/bf03040915.
  4. Truong PT, Parhar T, Hart J, et al. Population-based analysis of the frequency of anemia and its management before and during chemotherapy in patients with malignant lymphoma. Am J Clin Oncol. 2010;33(5):465–8. doi: 10.1097/coc.0b013e3181b4b147.
  5. Moullet I, Salles G, Ketterer N, et al. Frequency and significance of anemia in non-Hodgkin’s lymphoma patients. Ann Oncol. 1998;9:1109–15. doi: 10.1023/a:1008498705032.
  6. Samuelsson J. Long-standing resolution of anemia in symptomatic low-grade non-Hodgkin’s lymphoma patients treated with recombinant human erythropoietin as sole therapy. Med Oncol. 2002;19(1):69–72. doi: 10.1385/mo:19:1:69.
  7. Tsopra OA, Ziros PG, Lagadinou ED, et al. Disease-related anemia in chronic lymphocytic leukemia is not due to intrinsic defects of erythroid precursors: a possible pathogenetic role for tumor necrosis factor-alpha. Acta Haematol. 2009;121(4):187–95. doi: 10.1159/000220331.
  8. Eve HE, Rule SA. Autoimmune haemolytic anaemia associated with mantle cell lymphoma. Int J Hematol. 2010;91(2):322–5. doi: 10.1007/s12185-009-0489-9.
  9. Zent CS, Ding W, Reinalda MS, et al. Autoimmune cytopenia in chronic lymphocytic leukemia/small lymphocytic lymphoma: changes in clinical presentation and prognosis. Leuk Lymphoma. 2009;50(8):1261–8. doi: 10.1080/10428190903026492.
  10. Zupanic-Krmek D, Lang N, Jurcic D, et al. Analysis of the influence of various factors on anemia in patients with lymphoid malignancies. Acta Clin Croat. 2011;50(4):495–500.
  11. Романенко Н.А., Бессмельцев С.С., Розанова О.Е. и др. Влияние уровня ФНО-альфа на эффективность коррекции анемии у больных лимфопролиферативными заболеваниями. Онкогематология. 2010;3:22–8.
    [Romanenko NA, Bessmel’tsev SS, Rozanova OE, et al. Effect of TNF-alfa on efficacy of anemia management in patients with lymphoproliferative disorders. Onkogematologiya. 2010;3:22–8. (In Russ)]
  12. Рукавицын О.А. Актуальные вопросы диагностики и лечения анемии при хронических заболеваниях. Клиническая онкогематология. 2012;5(4):296–304.
    [Rukavitsyn OA. Urgent problems of diagnosis and treatment of anemia in chronic diseases. Klinicheskaya onkogematologiya. 2012;5(4):296–304. (In Russ)]
  13. Pierce CN, Larson DF. Inflammatory cytokine inhibition of erythropoiesis in patients implanted with a mechanical circulatory assist device. Perfusion. 2005;20(2):83–90. doi: 10.1191/0267659105pf793oa.
  14. Romanenko NA, Rozanova OE, Glazanova TV, Abdulkadyrov KM. Role of cytokines in resistance to erythropoiesis stimulating agents treatment of anaemia in patients with lymphoproliferative disorders. Haematologica. 2012;97(1):550–1.
  15. Лямкина А.С., Поспелова Т.И., Обгольц Ю.Н. и др. Патогенез и современная терапия анемического синдрома у пожилых больных лимфопролиферативными заболеваниями. Бюллетень СО РАМН. 2013;33(1):54–60.
    [Lyamkina AS, Pospelova TI, Obgol’ts YuN, et al. Pathogenesis and modern therapy of anemic syndrome in elderly patients with lymphoproliferative disorders. Byulleten’ SO RAMN. 2013;33(1):54–60. (In Russ)]
  16. Романенко Н.А., Бессмельцев С.С., Беркос М.В. и др. Прогностическая значимость ряда лабораторных показателей крови при использовании препаратов, стимулирующих эритропоэз у больных лимфопролиферативными заболеваниями с анемией. Терапевтический архив. 2013;85(8):81–6.
    [Romanenko NA, Bessmel’tsev SS, Berkos MV, et al. Prognostic relevance of several lab test blood parameters on the background of administration of erythropoiesis-stimulating agents in patients with lymphoproliferative disorders. Terapevticheskii arkhiv. 2013;85(8):81–6. (In Russ)]
  17. Gaya A, Urbano-Ispizua A, Fernandez-Aviles F, et al. Anemia associated with impaired erythropoietin secretion after allogeneic stem cell transplantation: incidence, risk factors, and response to treatment. Biol Blood Marrow Transplant. 2008;14(8):880–7. doi: 10.1016/j.bbmt.2008.05.008.
  18. Павлов А.Д., Морщакова Е.Ф., Румянцев А.Г. Эритропоэз, эритропоэтин, железо. М.: ГЭОТАР-Медиа, 2011. 304 с.
    [Pavlov AD, Morshchakova EF, Rumyantsev AG. Eritropoez, eritropoetin, zhelezo. (Erythropoiesis, erythropoietin, iron.) Moscow: GEOTAR-Media Publ.; 2011. 304 p. (In Russ)]
  19. Рукавицын О.А., Павлов А.Д. Анемии. СПб.: Д.-П., 2011. 240 с.
    [Rukavitsyn OA, Pavlov AD. Anemii. (Anemias.) Saint Petersburg: D.-P. Publ.; 2011. 240 p. (In Russ)]
  20. Цветаева Н.В., Левина А.А., Мамукова Ю.И. Основы регуляции обмена железа. Клиническая онкогематология. 2010;3(3):278–83.
    [Tsvetaeva NV, Levina AA, Mamukova YuI. Fundamental principles of iron metabliolism regulation. Klinicheskaya onkogematologiya. 2010;3(3):278–83. (In Russ)]
  21. Fleming RE, Sly WS. Ferroprotein mutation in autosomal dominant hemochromatosis: loss of function, gain in understanding. J Clin Inv. 2001;108:521–2. doi: 10.1172/jci13739.
  22. Сараева Н.О. Механизмы развития анемии при гемобластозах. Гематология и трансфузиология. 2007;52(1):31–7.
    [Saraeva NO. Mechanisms of development of anemia in hemoblastoses. Gematologiya i transfuziologiya. 2007;52(1):31–7. (In Russ)]
  23. Droge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002;82:47–95. doi: 10.1152/physrev.00018.2001.
  24. Романенко Н.А. Патогенез и коррекция анемического синдрома у больных лимфопролиферативными заболеваниями. Современная онкология. 2013;15(1):62–9.
    [Romanenko NA. Pathogenesis and therapy of anemic syndrome in patients with lymphoproliferative disorders. Sovremennaya onkologiya. 2013;15(1):62–9. (In Russ)]
  25. Mittelman M. The implications of anemia in multiple myeloma. Clin Lymphoma. 2003;4(1):23–9. doi: 10.3816/clm.2003.s.005.
  26. Han B, Shi YK, Zhu J, et al. Study on serum erythropoietin levels in patients with hematologic malignancies. Zhonghua Xue Ye Xue Za Zhi. 2006;27(8):543–5.
  27. Зубарева О.У., Клименко В.М. Повышение уровня провоспалительных цитокинов в раннем возрасте как фактор риска развития нервной и психической патологии. Российский физиологический журнал им. И.М. Сеченова. 2011;97(10):1048–59.
    [Zubareva OU, Klimenko VM. Increased levels of pro-inflammatory cytokines at an early age as a risk factor of neurological and mental disorders. Rossiiskii fiziologicheskii zhurnal im. IM. Sechenova. 2011;97(10):1048–59. (In Russ)]
  28. Клименко В.М., Зубарева О.Е., Краснова И.Н. Роль внутримозговых рецепторов интерлейкина-1 в модуляции гомеостатических реакций организма. Нейрохимия. 1995;12(2):16–22.
    [Klimenko VM, Zubareva OE, Krasnova IN. Role of intracerebral interleukin-1 receptors in modulation of homeostatic reactions of body. Neirokhimiya. 1995;12(2):16–22. (In Russ)]
  29. Wang T, Tu MF, Zhu J. The role of cytokines in lymphoma with anemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2013;21(2):392–5.
  30. Поспелова Т.И., Лямкина А.С. Уровень цитокинов (интерлейкина-1b, фактора некроза опухолей-a, интерферона-g, интерлейкина-6) у больных лимфопролиферативными заболеваниями с анемическим синдромом. Анемия при лимфомах: научное издание. Новосибирск: НГМУ, 2008. С. 97–114.
    [Pospelova TI, Lyamkina AS. Uroven’ tsitokinov (interleikina-1b, faktora nekroza opukholei-a, interferona-g, interleikina-6) u bol’nykh limfoproliferativnymi zabolevaniyami s anemicheskim sindromom. Anemiya pri limfomakh: nauchnoe izdanie. (Levels of cytokines (interleukin-1b, tumor necrosis factor-a, interferon-g, and interleukin-6) in LPD patients with anemic syndrome. Anemia in lymphomas: scientific publication.) Novosibirsk: NGMU Publ.; 2008. p. 97–114. (In Russ)]
  31. Hohaus S, Massini G, Giachelia M, et al. Anemia in Hodgkin’s lymphoma: the role of interleukin-6 and hepcidin. J Clin Oncol. 2010;28(15):2538–43. doi: 10.1200/jco.2009.27.6873.
  32. Романенко Н.А. Патогенез и терапия анемии препаратами рекомбинантного эритропоэтина у онкогематологических больных (обзор литературы). Онкогематология. 2012;3:20–9.
    [Romanenko NA. Pathogenesis and therapy of anemia using recombinant erythropoietin agents in oncohematological patients (literature review). Onkogematologiya. 2012;3:20–9. (In Russ)]
  33. Gans T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood. 2003;102(3):783–90. doi: 10.1182/blood-2003-03-0672.
  34. Nemeth E, Rivera S, Gabajan V, et al. IL-6 mediates hyppoferremia inducting the synthesis of the iron regulatory hormone hepcidin. J Clin Inv. 2004;113:1271–6. doi: 10.1172/jci200420945.
  35. Pigeon C, Ilyin G, Courselaud B, et al. A new mouse liver specific protein homologous to human antibacterial peptid hepcidin is overexpressed during iron overload. J Biol Hem. 2001;276:7811–9. doi: 10.1074/jbc.m008923200.
  36. Buck I, Morceau F, Cristofanon S, et al. Tumor necrosis factor alpha inhibits erythroid differentiation in human erythropoietin-dependent cells involving p38 MAPK pathway, GATA-1 and FOG-1 down regulation and GATA-2 up regulation. Biochem Pharmacol. 2008;76(10):1229–39. doi: 10.1016/j.bcp.2008.08.025.
  37. Грицаев С.В., Даваасамбуу Б., Романенко Н.А., Абдулкадыров К.М. Отбор больных для терапии хелаторами железа. Клиническая онкогематология. 2013;6(2):204–9.
    [Gritsaev SV, Davaasambuu B, Romanenko NA, Abdulkadyrov KM. Selection of patients for treatment with iron chelators. Klinicheskaya onkogematologiya. 2013;6(2):204–9. (In Russ)]
  38. Птушкин В.В. Дискуссионные вопросы применения эритропоэтинов в лечении анемии у пациентов с опухолевыми заболеваниями. Онкогематология. 2007;2:31–6.
    [Ptushkin VV. Depatable questions of applying erythropoietin in treatment of anemic patients with tumors. Onkogematologiya. 2007;2:31–6. (In Russ)]
  39. Трансфузиология: национальное руководство. Под ред. А.А. Рагимова. М.: ГЭОТАР-Медиа, 2012. 1184 с.
    [Ragimov AA, ed. Transfuziologiya: natsional’noe rukovodstvo. (Transfusiology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2012. 1184 p. (In Russ)]
  40. Романенко Н.А. Коррекция и лечение анемии у больных с гемобластозами эритроцитсодержащими компонентами крови и препаратами рекомбинантного эритропоэтина. Вестник гематологии. 2007;3(4):46–54.
    [Romanenko NA. Correction and treatment of anemia in patients with hemoblastoses using RBC-containing blood components and recombinant erythropoietin agents. Vestnik gematologii. 2007;3(4):46–54. (In Russ)]
  41. Техническое руководство американской ассоциации банков крови. Под ред. Ю.Н. Токарева. Милан: Европейская школа трансфузионной медицины, 2000. 1056 с.
    [Tokarev YuN, ed. Tekhnicheskoe rukovodstvo amerikanskoi assotsiatsii bankov krovi. (Technical manual of the American Association of Blood Banks.) Milan: European School of Transfusion Medicine Publ.; 2000. 1056 p. (In Russ)]
  42. Aapro MS, Jelkmann W, Constantinescu SN, Leyland-Jones B. Effects of erythropoietin receptors and erythropoiesis-stimulating agents on disease progression in cancer. Br J Cancer. 2012;106(7):1249–58. doi: 10.1038/bjc.2012.42.
  43. Brecher ME, Hay SN. Bacterial contamination of blood components. Clin Microbiol Rev. 2005;18(1):195–204. doi: 10.1128/cmr.18.1.195-204.2005.
  44. Khorana AA, Francis CW, Blumberg N, et al. Blood Transfusions, Thrombosis, and Mortality in Hospitalized Patients With Cancer. Arch Intern Med. 2008;168(21):2377–81. doi: 10.1001/archinte.168.21.2377.
  45. Klein HG, Spahn DR, Carson JL. Red Blood Cell Transfusion in clinical practice. The Lancet. 2007;370:415–26. doi: 10.1016/s0140-6736(07)61197-0.
  46. Leitch HA, Vickars LM. Supportive care and chelation therapy in MDS: are we saving lives or just lowering iron? Hematol Am Soc Hematol Educ Program. 2009:664–72. doi: 10.1182/asheducation-2009.1.664.
  47. Жибурт Е.Б., Караваев А.В., Шестаков Е.А. и др. Правила переливания эритроцитов, основанные на доказательствах. Трансфузиология. 2012;13(3):55.
    [Zhiburt EB, Karavaev AV, Shestakov EA, et al. Evidence-based guidelines for RBC transfusions. Transfuziologiya. 2012;13(3):55. (In Russ)]
  48. Romanenko N, Kostroma I, Bulgan D, et al. Analysis of Quality-of-Life after Red Blood Cell Transfusions in Hematological Malignancies Patients with Anaemia. Haematologica. 2014;99(1):787.
  49. Романенко Н.А., Беркос М.В., Бессмельцев С.С. и др. Прогностическое значение сывороточного эритропоэтина при коррекции анемии препаратами рекомбинантного эритропоэтина у пациентов с лимфопролиферативными заболеваниями. Казанский медицинский журнал. 2012;93(4):584–90.
    [Romanenko NA, Berkos MV, Bessmel’tsev SS, et al. Prognostic significance of serum erythropoietin in correction of anemia in patients with lymphoproliferative disorders using recombinant erythropoietin. Kazanskii meditsinskii zhurnal. 2012;93(4):584–90. (In Russ)]
  50. Henry DH. Guidelines and recommendations for the management of anemia in patients with lymphoid malignancies. Drugs. 2007;67(2):175–94. doi: 10.2165/00003495-200767020-00002.
  51. Rizzo JD, Brouwers M, Hurley P, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood. 2010;116(20):4045–59. doi: 10.1182/blood-2010-08-300541.
  52. Haioun C, Salar A, Pettengell R, et al. Anemia and erythropoiesis-stimulating agent administration in patients with non-Hodgkin lymphoma treated with cyclophosphamide, doxorubicin, vincristine, and prednisolone with/without rituximab chemotherapy: results from an observational study. Leuk Lymphoma. 2011;52:796–803. doi: 10.3109/10428194.2011.557166.
  53. Romanenko N, Kostroma I, Karmatskaya I, Potikhonova N. Improvement of quality of life and increase hemoglobin concentration in anemic patients with lymphoproliferative disorders treated erythropoiesis-stimulating agents. Haematologica. 2013;98(1):756.
  54. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома. Современный взгляд на проблему. Алматы, 2007. 480 с.
    [Bessmel’tsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma. Sovremennyi vzglyad na problemu. (Multiple myeloma. Modern view on the problem.) Almaty; 2007. 480 p. (In Russ)]
  55. Christodoulou C, Dafni U, Aravantinos G, et al. Effects of Epoetin-a on Quality of Life of Cancer Patients with Solid Tumors Receiving Chemotherapy. Anticancer Res. 2009;29:693–702.
  56. Yoshimura A, Kobayashi K, Fumimoto H, et al. Cross-Cultural Validation of Japanese Functional Assessment of Cancer-Therapy Anemia (FACT-An). J Nippon Med Sch. 2004;71(5):314–22. doi: 10.1272/jnms.71.314.
  57. Campos MPO, Hassan BJ, Riechelmann R, Del Giglio A. Cancer-related fatigue: a practical review. Ann Oncol. 2011;22(6):1273–9. doi: 10.1093/annonc/mdq458.
  58. Cella D. The Functional Assessment of Cancer Therapy-Anemia (FACT-An) Scale: a new tool for the assessment of outcomes in cancer anemia and fatigue. Semin Hematol. 1997;34(3–2):13–9.
  59. Yellen SB, Cella DF, Webster K, et al. Measuring fatigue, and other anemia related symptoms with the Functional Assessment of Cancer Therapy (FACT) measurement system. J Pain Symptom Manage. 1997;13:63–74. doi: 10.1016/s0885-3924(96)00274-6.
  60. Романенко Н.А., Бессмельцев С.С., Кармацкая И.И. и др. Эффективность эритропоэзстимулирующих препаратов и их влияние на качество жизни больных с анемией при лимфопролиферативных заболеваниях. Казанский медицинский журнал. 2013;94(4):468–73.
    [Romanenko NA, Bessmel’tsev SS, Karmatskaya II, et al. Efficacy of erythropoiesis-stimulating agents and their impact on the quality of life of anemic patients with lymphoproliferative disorders. Kazanskii meditsinskii zhurnal. 2013;94(4):468–73. (In Russ)]
  61. Аппалуп М.В., Майорова О.А. Посттрансфузионные осложнения: с чего начинается Haemovigilance. Трансфузиология. 2014;15(3):31–6.
    [Appalup MV, Maiorova OA. Post-transfusion complications: what Haemovigilance begins with. Transfuziologiya. 2014;15(3):31–6. (In Russ)]
  62. Приказ от 2 апреля 3013 г. № 183 н «Об утверждении правил клинического использования донорской крови и (или) ее компонентов» [электронный документ]. Доступно по: http://www.transfusion.ru/2013/08-29-1.pdf.
    [Decree No. 183 н dated April 2, 2013, On approval of guidelines for clinical use of donor blood and/or its components [Internet]. Available from: http://www.transfusion.ru/2013/08-29-1.pdf. (In Russ)]
  63. Ярославцева Н.Г., Грумбкова Л.О., Туполева Т.А. и др. Вирусная безопасность гемотрансфузий: обеспечивают ли ее принятые лабораторные методы выбраковки донорской крови по гепатитам В и С. Гематология и трансфузиология. 2006;51(2):22–6.
    [Yaroslavtseva NG, Grumbkova LO, Tupoleva TA, et al. Viral safety of hemotransfusion: do known lab tests used for rejection of donor blood due to hepatitis B and C ensure it? Gematologiya i transfuziologiya. 2006;51(2):22–6. (In Russ)]
  64. Benjamin RJ. Bacterial contamination. Vox Sanguinis. 2013;105(Suppl. 2):3.

 

Anemias and iron deficiency in cancer patients

V.V. Ptushkin

Federal Scientific and Clinical Centre of Pediatric Hematology, Oncology and Immunology named after Dmitriy Rogachev, Moscow, Russian Federation


ABSTRACT

Anemia is frequent in cancer patients and its incidence increases with chemotherapy. Anemia negatively impacts survival and accentuates fatigue in cancer patients. Cancer promotes inflammatory cytokine production, which suppresses erythropoiesis and erythropoietin production. Erythropoiesis-stimulating agents improve erythropoiesis and reduce transfusion needs in anemic cancer patients receiving chemotherapy. However, meta-analyses have shown an increased risk of thromboembolic events with еrythropoiesis-stimulating agents use during chemotherapy, but not increased on-study mortality or reduced overall survival. Inflammatory cytokine production in patients with cancer, reduce the availability of iron for effective erythropoiesis. This review summarises clinical consequences of iron deficiency and anaemia in cancer patients, mechanisms how impaired iron homeostasis affects diagnosis and treatment of iron deficiency, and data from clinical trials evaluating i.v. iron with or without concomitant erythropoiesis-stimulating agents.


Keywords: Anemia, cancer, erythropoietin, iron propagates, ferric carboxymaltose, ferritin, transferrin

Read in PDF (RUS)pdficon


REFERENCES

  1. Ludwig H., Van B.S., Barrett-Lee P. et al. The European Cancer AnaemiaSurvey (ECAS): a large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur. J. Cancer 2004; 40: 2293–306.
  2. Groopman J.E., Itri L.M. Chemotherapy-induced anemia in adults: incidence and treatment. J. Natl. Cancer Inst. 1999; 91: 1616–34.
  3. Beale A.L., Penney M.D., Allison M.C. The prevalence of iron deficiency among patients presenting with colorectal cancer. Colorectal. Dis. 2005; 7: 398–402.
  4. Kuvibidila S.R., Gauthier T., Rayford W. Serum ferritin levels and transferrin saturation in men with prostate cancer. J. Natl. Med. Assoc. 2004; 96: 641–9.
  5. Steinmetz H.T., Tsamaloukas A., Schmitz S. et al. A new concept for the differential diagnosis and therapy of anaemia in cancer patients. Support Care Cancer 2010; 19: 261–9.
  6. Beguin Y., Lybaert W., Bosly A. A prospective observational study exploring the impact of iron status on response to darbepoetin alfa in patients with chemotherapy induced anemia. Blood 2009; 114 (Abstr 2007).
  7. Ludwig H., Muldur E., Endler G. et al. High prevalence of iron deficiency across different tumors correlates with anemia, increases during cancer treatment and is associated with poor performance status. Haematologica 2011; 96 (Abstr 982).
  8. Anker S.D., Comin C.J., Filippatos G. et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N. Engl. J. Med. 2009; 361: 2436–48.
  9. Crawford J., Cella D., Cleeland C.S. et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002; 95: 888–95.
  10. Auerbach M., Ballard H., Trout J.R. et al. Intravenous iron optimizes the response to recombinant human erythropoietin in cancer patients with chemotherapy-related anemia: a multicenter, open-label, randomized trial. J. Clin. Oncol. 2004; 22: 1301–7.
  11. Rizzo J.D., Brouwers M., Hurley P. et al. American Society of Hematology/ American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood 2010; 116: 4045–59.
  12. Auerbach M., Silberstein P.T., Webb R.T. et al. Darbepoetin alfa 300 or 500 mg once every 3 weeks with or without intravenous iron in patients with chemotherapyinduced anemia. Am. J. Hematol. 2010; 85: 655–63.
  13. Henry D.H., Dahl N.V., Auerbach M. et al. Intravenous ferric gluconate significantly improves response to epoetin alfa versus oral iron or no iron in anemic patients with cancer receiving chemotherapy. Oncologist 2007; 12: 231–42.
  14. Pedrazzoli P., Farris A., Del P.S. et al. Randomized trial of intravenous iron supplementation in patients with chemotherapy-related anemia without iron deficiency treated with darbepoetin alpha. J. Clin. Oncol. 2008; 26: 1619–25.
  15. Beguin Y. Prediction of response and other improvements on the limitations of recombinant human erythropoietin therapy in anemic cancer patients. Haematologica 2002; 87: 1209–21.
  16. Wish J.B. Assessing iron status: beyond serum ferritin and transferrin saturation. Clin. J. Am. Soc. Nephrol. 2006; 1(Suppl. 1): S4–S8.
  17. Hedenus M., Birgegard G., Nasman P et al. Addition of intravenous iron to epoetin beta increases hemoglobin response and decreases epoetin dose requirement in anemic patients with lymphoproliferative malignancies: a randomized multicenter study. Leukemia 2007; 21: 627–32.
  18. Ludwig H., Endler G., Hubl W. et al. High prevalence of iron deficiency in patients with various hematological and malignant diseases: a single center study in 1989 sequential patients. Haematologica 2010; 95 (Abstr 1819).
  19. Ludwig H., Aapro M., Bokemeyer C. et al. Treatment patterns and outcomes in the management of anaemia in cancer patients in Europe: findings from the Anaemia Cancer Treatment (ACT) study. Eur. J. Cancer 2009; 45: 1603–15.
  20. Aapro M.S., Link H. September 2007 update on EORTC guidelines and anemia management with erythropoiesis-stimulating agents. Oncologist 2008; 13(Suppl. 3): 33–6.
  21. Vamvakas E.C., Blajchman M.A. Transfusion-related mortality: the ongoing risks of allogeneic blood transfusion and the available strategies for their prevention. Blood 2009; 113: 3406–17.
  22. Marik P.E., Corwin H.L. Efficacy of red blood cell transfusion in the critically ill: a systematic review of the literature. Crit. Care Med. 2008; 36: 2667–74.
  23. Thomson A., Farmer S., Hofmann A. et al. Patient blood management—a new paradigm for transfusion medicine? ISBT Sci. Series 2009; 4: 423–35.
  24. Amato A.C., Pescatori M. Effect of perioperative blood transfusions on recurrence of colorectal cancer: meta-analysis stratified on risk factors. Dis. Colon Rectum 1998; 41: 570–85.
  25. Bohlius J., Schmidlin K., Brillant C. et al. Erythropoietin or darbepoetin for patients with cancer—meta-analysis based on individual patient data. Cochrane Database Syst. Rev. 2009; CD007303.
  26. Gabrilove J.L., Cleeland C.S., Livingston R.B. et al. Clinical evaluation of once-weekly dosing of epoetin alfa in chemotherapy patients: improvements in hemoglobin and quality of life are similar to three-times-weekly dosing. J. Clin. Oncol. 2001; 19: 2875–82.
  27. Littlewood T.J., Bajetta E., Nortier J.W. et al. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: results of a randomized, double-blind, placebo-controlled trial. J. Clin. Oncol. 2001; 19: 2865–74.
  28. US Food and Drug Administration. Epoetin Alfa (Marketed as Epoetin, Procrit) Label. http://www.accessdata.fda.gov/drugsatfda_docs/ label/2010/103234s5199lbl.pdf (5 September 2011, date last accessed).
  29. US Food and Drug Administration. Darbepoetin Alfa (Marketed as Aransep) Label. http://www.accessdata.fda.gov/drugsatfda_docs/ label/2010/103951s5197lbl.pdf (5 September 2011, date last accessed).
  30. Macdougall I.C. Strategies for iron supplementation: oral versus intravenous. Kidney Int. Suppl. 1999; 69: S61–S66.
  31. Bastit L., Vandebroek A., Altintas S. et al. Randomized, multicenter, controlled trial comparing the efficacy and safety of darbepoetin alpha administered every 3 weeks with or without intravenous iron in patients with chemotherapyinduced anemia. J. Clin. Oncol. 2008; 26: 1611–8.
  32. Steensma D.P., Sloan J.A., Dakhil S.R. et al. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy associated anemia. J. Clin. Oncol. 2011; 29: 97–105.
  33. Gafter-Gvili A., Rozen-Zvi B., Vidal L. et al. Intravenous iron supplementation for the treatment of cancer-related anemia—systematic review and metaanalysis. Blood 2010; 116 (Abstr 4249).
  34. Petrelli F., Borgonovo K., Cabiddu M. et al. Addition of iron to erythropoiesis-stimulating agents in cancer patients: a meta-analysis of randomized trials. J. Cancer Res. Clin. Oncol. 2012; 138: 179–87.
  35. Dangsuwan P., Manchana T. Blood transfusion reduction with intravenous iron in gynecologic cancer patients receiving chemotherapy. Gynecol. Oncol. 2010; 116: 522–5.
  36. Steinmetz T., Tschechne B., Virgin G. et al. Ferric carboxymaltose for the correction of cancer- and chemotherapy-associated anemia in clinical practice. Haematologica 2011; 96 (Abstr 983).
  37. Evstatiev R., Marteau P., Iqbal T. et al. FERGIcor, a randomized controlled trial on ferric carboxymaltose for iron deficiency anemia in inflammatory bowel disease. Gastroenterology 2011; 141: 846–53.
  38. Kulnigg S., Stoinov S., Simanenkov V. et al. A novel intravenous iron formulation for treatment of anemia in inflammatory bowel disease: the ferric carboxymaltose (FERINJECT) randomized controlled trial. Am. J. Gastroenterol. 2008; 103: 1182–92.
  39. Aapro M., Beguin Y., Birgegard G. et al. Too-low iron doses and too many dropouts in negative iron trial? J. Clin. Oncol. 2011; 29: e525–e526.
  40. Bailie G.R., Horl W.H., Verhof J.J. Differences in spontaneously reported hypersensitivity and serious adverse events for intravenous iron preparations: comparison of Europe and North America. Drug Res. 2011; 61: 267–75.
  41. Bailie G.R., Clark J.A., Lane C.E. et al. Hypersensitivity reactions and deaths associated with intravenous iron preparations. Nephrol. Dial. Transplant. 2005; 20: 1443–9.
  42. Chertow G.M., Mason P.D., Vaage-Nilsen O. et al. Update on adverse drug events associated with parenteral iron. Nephrol. Dial. Transplant. 2006; 21: 378–82.
  43. Zhang F., Wang W., Tsuji Y. et al. Post-transcriptional modulation of iron homeostasis during p53-dependent growth arrest. J. Biol. Chem. 2008; 283: 33911–8.
  44. Baliga R., Zhang Z., Baliga M. et al. In vitro and in vivo evidence suggesting a role for iron in cisplatin-induced nephrotoxicity. Kidney Int. 1998; 53: 394–401.
  45. Toyokuni S. Role of iron in carcinogenesis: cancer as a ferrotoxic disease. Cancer Sci. 2009; 100: 9–16.
  46. Bergeron R.J., Streiff R.R., Elliott G.T. Influence of iron on in vivo proliferation and lethality of L1210 cells. J. Nutr. 1985; 115: 369–74.
  47. Carthew P., Nolan B.M., Smith A.G. et al. Iron promotes DEN initiated GST-P foci in rat liver. Carcinogenesis 1997; 18: 599–603.
  48. Auerbach M., Glaspy J. What is the right balance between iron and erythropoiesis stimulating agents in chemotherapy induced anemia? Eur. J. Clin. Med. Oncol. 2009; 1: 7–12.
  49. Beguin Y., Maertens J., De Prijck B. et al. Darbepoetin-alfa and i.v. iron administration after autologous hematopoietic stem cell transplantation: a prospective randomized multicenter trial. Blood 2008; 112 (Abstr 54).